Vous êtes sur la page 1sur 7

Review Article

Transfus Med Hemother 2015;42:287293


DOI: 10.1159/000439002

Received: May 29, 2015


Accepted: July 28, 2015
Published online: September 7, 2015

New Insights in the Pathogenesis of Autoimmune


Hemolytic Anemia
Wilma Barcellini
U.O. Oncoematologia, Fondazione IRCCS Ca Granda Ospedale Maggiore Policlinico, Milan, Italy

Keywords
Autoimmune hemolytic anemia Autoantibodies
Cytokines
Summary
Autoimmune hemolytic anemia (AIHA) is caused by the
increased destruction of red blood cells (RBCs) by antiRBC autoantibodies with or without complement activation. RBC destruction may occur both by a direct lysis
through the sequential activation of the final components of the complement cascade (membrane attack
complex), or by antibody-dependent cell-mediated cytotoxicity (ADCC). The pathogenic role of autoantibodies
depends on their class (the most frequent are IgG and
IgM), subclass, thermal amplitude (warm and cold
forms),as well as affinity and efficiency in activating
complement. Several cytokines and cytotoxic mechanisms (CD8+ T and natural killer cells) are further involved in RBC destruction. Moreover, activated macrophages carrying Fc receptors may recognize and
phagocyte erythrocytes opsonized by autoantibodies
and complement. Direct complement-mediated lysis
takes place mainly in the circulations and liver, whereas
ADCC, cytotoxicity, and phagocytosis occur preferentially in the spleen and lymphoid organs. The degree of
intravascular hemolysis is 10-fold greater than extravascular one. Finally, the efficacy of the erythroblastic compensatory response can greatly influence the clinical picture of AIHA. The interplay and relative burden of all
these pathogenic mechanisms give reason for the great
clinical heterogeneity of AIHAs, from fully compensated
to rapidly evolving fatal cases.
2015 S. Karger GmbH, Freiburg

2015 S. Karger GmbH, Freiburg


16603796/15/04250287$39.50/0
Fax +49 761 4 52 07 14
Information@Karger.com
www.karger.com

Accessible online at:


www.karger.com/tmh

Introduction
Autoimmune hemolytic anemia (AIHA) is defined as the increased destruction of red blood cells (RBCs) in the presence of
anti-RBC autoantibodies with or without complement activation.
Autoantibodies are produced by both tissue and circulating selfreactive B lymphocytes, following cooperation with T helper lymphocytes. A third key player in anti-RBC autoimmunity is the
complement system, which is able to induce a direct erythrocyte
osmotic lysis through the sequential activation of the membrane
attack complex (MAC). Moreover, autoantibodies are able to destroy RBCs by antibody-dependent cell-mediated cytotoxicity
(ADCC), which is mediated by cytotoxic CD8+ T cells and natural
killer (NK) that carry membrane receptors for the Fc portion of
immunoglobulin G (IgG). Finally, activated macrophages carrying
Fc receptors may recognize and phagocyte erythrocytes opsonized
by autoantibodies and complement. While direct complementmediated lysis takes place mainly in the circulations and liver,
ADCC and phagocytosis occur preferentially in the spleen and
lymphoid organs [1].
AIHAs can be distinguished on the basis of the autoantibody Ig
class and thermal characteristics, i.e. the optimal temperature of reaction with autologous erythrocytes, in warm (wAIHA), cold (cold
agglutinin disease; CAD), and mixed forms, although atypical cases
of difficult diagnostic classification are reported with increasing
frequency [13]. Moreover, AIHAs are classified in primary (idiopathic), in which hemolysis dominates the clinical picture in the
absence of any other coexisting disorder, and in secondary forms,
accompanying and complicating an underlying disease. The majority of warm forms are idiopathic or associated with other autoimmune diseases; acute cold forms are mainly secondary to infections, whereas chronic CADs are frequently associated to lymphoproliferative or neoplastic diseases [13]. All these factors account for the great clinical heterogeneity of AIHAs, from
compensated forms without anemia to fulminating disease. Table1
shows the main characteristics of AIHAs.

Dr. Wilma Barcellini


U.O. Oncoematologia, Padiglione Granelli
Fondazione IRCCS Ca Granda Ospedale Maggiore Policlinico
Via F. Sforza,35, 20122 Milan, Italy
wbarcel@policlinico.mi.it

It is commonly thought that autoimmunity is the result of the


interaction of genetic predisposition and environmental factors.
While several HLA associations have been reported for various
organ and systemic autoimmune diseases (type-1 insulin-dependent diabetes, Hashimoto thyroiditis, Graves disease, multiple sclerosis, myasthenia gravis, pemphigus vulgaris, systemic lupus erythematosus, rheumatoid arthritis and ankylosing spondylitis), no
clear HLA linkage has been found for AIHA. Moreover, the mechanisms involved in the breakdown of both central and peripheral
tolerance in the development of AIHA are still largely unknown.
Central tolerance occurs during the early differentiation of B cells
in the bone marrow and T cells in the tymus, where autoreactive
cells undergo apoptosis upon binding to self-antigens (negative selection). Peripheral tolerance is an active process, where anergy or
suppression against self-antigens is driven by specific CD4+/
CD25+ regulatory T cells (Tregs) and CD8+ suppressor T lymphocytes [4].
Molecular mimicry between self-antigens and foreign antigens
is one of several mechanisms by which pathogens might be involved in the initiation and/or acceleration of autoimmunity. This
is likely the prevalent mechanism of AIHAs secondary to various
bacterial, mycoplasma or virus infections, probably due to the
presence of common antigenic epitopes on RBC proteins or carbohydrates. Additional mechanisms involved in the failure to maintain self-tolerance are polyclonal lymphocyte activation, again induced by several viral infections, and the emergence of forbidden
clones, as observed in lymphoproliferative diseases. This hypothesis, proposed more than 60 years ago by Burnet and based on the
persistence of self-reactive clones of lymphocytes that should have
been deleted via immune tolerance, has gained new interest due to
the success of B-cell-depleting therapy in several autoimmune diseases [5]. A particular condition is related to the generation of neoantigens by environmental agents or drugs, as observed in druginduced AIHA, where the autoimmune response persists until they
are destroyed and eliminated [6, 7]. Moreover, in a large study of

Table 1. Classification of AIHAs

717 patients displaying anti-RBC autoantibodies, about one-third


was found positive for alloantibodies, and RBC transfusion was
identified as a major cause for autoimmunization [8]. Recently, attention has been focused on the autoimmune/inflammatory syndrome induced by adjuvants, which include several still undefined
diseases, such as siliconosis, Gulf War syndrome, Spanish toxic oil
syndrome, Ardystil syndrome, and post-vaccination [9]. Above all
there is a loss of suppressor mechanisms and the alteration of the
regulatory cells and cytokine network involved in the control of
self-tolerance [4].

Autoantibodies in the Pathogenesis of AIHA


The most frequent autoantibodies against RBCs are IgG, which
mainly determine extravascular hemolysis through the ADCC in
the reticuloendothelial system (spleen and to a lesser extent liver).
The IgG subclass influences the degree to which these antibodies
shorten RBC survival: IgG1 is the most commonly encountered
subclass and together with IgG3 shortens the half-life more efficiently than IgG2 and IgG4. Autoantibodies of this class are mostly
directed against epitopes of the Rh system. They generally react at
37 C and, therefore, are responsible for the warm forms of AIHA
[1]. IgG are able to activate the complement cascade, with the exception of IgG2 and IgG4 subclasses. IgA autoantibodies are generally associated with IgG and only rarely reported as the sole causative agent of AIHA.
IgM are pentameric autoantibodies able to dramatically fix
complement compared with other isotypes, thus prevalently causing intravascular hemolysis, and to a lesser extent C3d-mediated
extravascular lysis (mainly in the liver). Autoantibodies of this class
are directed against the I/i system. Their optimal temperature of
reaction is 4 C, and thus they are responsible for the cold forms of
AIHA [1, 2]. However, the thermal amplitude of IgM autoantibodies ranges from 0 to 34 C, and those with a thermal activity close

1) Warm autoimmune hemolytic anemia (wAIHA)


DAT positive for IgG or IgG+C3d
IgG antibody directed mainly against epitopes of the Rh system
Idiopathic or secondary to autoimmune, lymphoproliferative, or neoplastic diseases
2) Cold autoimmune hemolytic anemia
2a) Cold agglutinin disease (CAD)
DAT positive for C3d
IgM directed against the I/i system
Idiopathic or secondary to infections (acute transitory) or to lymphoproliferative diseases (chronic)
2b) Paroxysmal cold hemoglobinuria (PCH)
Donath-Landsteiner test positive
Bithermic IgG directed against the erythrocyte P antigen
Idiopathic or secondary to syphilis (chronic) or to other infections (acute transitory)
3) Mixed autoimmune hemolytic anemia
DAT positive for IgG and C3d
Coexistence of warm IgG autoantibodies and high titer IgM cold agglutinins
Idiopathic or secondary to lymphoproliferative or autoimmune diseases

288

Transfus Med Hemother 2015;42:287293

Barcellini

to physiological temperatures (warm IgM) are the most harmful


autoantibodies, able to cause severe forms of AIHA with a reported
mortality rate of about 20% [6]. In fact, it is worth reminding that
the amount of RBC destruction by intravascular hemolysis has
been calculated in 200 ml of RBCs in 1 h, whereas by extravascular
hemolysis it is 10-fold lesser (0.25 ml RBCs/kg/h, i.e., for a patient
weighing 70 kg = 17.5 ml RBCs/h = 420 ml RBCs / 24 h) [1, 2].
Anti-RBC antibodies are usually detected by the direct antiglobulin test (DAT) or Coombs test, generally performed by the
traditional tube agglutination technique with polyspecific antisera.
Nowadays the use of monospecific anti-IgG, anti-IgM, and antiC3 antisera is recommended to define the class of the autoantibody and its thermal amplitude. It is worth mentioning a simple
test, the spontaneous agglutination of RBCs at 20 C, a characteristic feature indicating the presence of cold IgM autoantibodies.
According to DAT results and to the thermal characteristics of the
autoantibody, AIHAs are usually classified in two main groups:
wAIHA (70% of cases), which displayed IgG only or IgG plus
C3d on erythrocytes, and CAD (20% of patients), which showed
C3d only, along with high serum titer of cold agglutinins of I specificity. There are few cases (78% of all AIHAs), which showed a
DAT positive for IgG and C3d, with the coexistence of warm autoantibodies and high-titer cold agglutinins (mixed AIHA). However, there is increasing evidence of atypical cases of difficult classification mainly DAT-negative which are frequently severe
and refractory/relapsed after several therapy lines, and may have a
fatal outcome [10, 11]. DAT-negative cases may be caused by
warm IgM autoantibodies, which can be identified by the dual direct antiglobulin test (DDAT) [12, 13]. It is worth mentioning the
Donath-Landsteiner autoantibody, a bithermic hemolysin able to
fix complement at cold temperatures and to determine RBCs lysis
at 37 C, that is directed against the erythrocyte P antigen and responsible of paroxysmal cold hemoglobinuria, which is rather
common in children (30% of cases) but rare in adults (<1% of all
AIHA cases) [1, 2].
Finally, DAT positivity may also be due to drug-induced antibodies that are associated with a variety of drugs (antimicrobials:
42% of cases, anti-inflammatory drugs: 15%, and anti-neoplastics:
11%) but occur rarely (about 1 in 1 million of the population).
Drug-dependent antibodies react in vitro only in the presence of
drug bound to RBCs or added to the patients serum in test systems
(typical example penicillin and cefotetan). Drug-independent antibodies can be detected in vitro without adding any drug, thus with
characteristics identical to classic autoantibodies. The typical example is methyldopa, which causes the production of RBC autoantibodies in about 15% of the patients receiving the drug, but only
about 1% develops AIHA [1, 6, 7, 14, 15].

The Role of Complement in the Pathogenesis of


AIHA
The complement system plays a role in both warm AIHA and
CAD. In the former cases IgG bound to erythrocytes are able to fix

New Insights in the Pathogenesis of Autoimmune


Hemolytic Anemia

complement protein complex C1 and thus activate the classical


complement pathway; IgG3 subclass is the more efficient, followed
by IgG1, whereas IgG2 and IgG4 are less or not active. Warm IgM
autoantibodies are potent complement activators but they are usually not found on RBCs. In fact, they temporary bind, activate complement, and then detach from the membrane, where only complement factors may be demonstrated. Following complement activation, phagocytosis of C3b-opsonized erythrocytes occurs mainly in
the liver, although complete intravascular hemolysis mediated by
the terminal lytic complex may occur as well [12, 1618]. DAT
positivity for C3 fragments is a marker of complement involvement in wAIHA.
The role of the complement system in CAD is also significant,
provided that the thermal amplitude of the IgM autoantibody is
close to values found on the surface of the body areas exposed to
external cold temperatures. IgM-cold autoantibody binds C1 and
thereby initiates the classical complement pathway [19]: C1 esterase activates C4 and C2, generating C3 convertase, which results in
the cleavage of C3 to C3a and C3b. When RBCs return to the central parts of the body with a temperature of 37 C, the IgM autoantibody detaches from the cell surface, while C3b remains bound. A
part of the C3b-coated RBCs is sequestered in the liver, whereas the
other erythrocytes are lysed through the activation of C5 by C3b.
The final formation of the MAC determines intravascular hemolysis [2022]. This occurs in severe acute exacerbations and in some
severe hemolytic patients, as evidenced by the finding of hemoglobinuria, a typical finding of paroxysmal nocturnal hemoglobinuria
(PNH). This pathogenic mechanism accounts for the beneficial effect of C5 inhibition with the monoclonal antibody eculizumab in
some cases [23, 24]. Further evidence for the potential use of complement inhibitors in the treatment of AIHA is the finding that
TNT003, a mouse monoclonal antibody targeting complement
protein C1s, prevents induction of in vitro hemolysis by cold agglutinins [25]. Moreover, a C1-esterase inhibitor concentrate was
found to protect erythrocytes from complement-mediated destruction in AIHA [26]. Further complement inhibitors, such as compstatin Cp40, a low-molecular-peptide that prevents cleavage of C3,
and TT30, which prevents C3 convertase activation, have shown
promising effects on complement-mediated lysis of erythrocytes in
PNH [27, 28], and may be potential treatments suitable for acute
hemolysis in AIHA.
It is worth reminding that cold IgM antibodies are frequently
monoclonal and, in more than 90% of the patients, show light
chain restriction [29]. Moreover, CAD patients have frequently an
associated clonal B-cell lymphoproliferative disorder (lymphoplasmacytic lymphoma, marginal zone lymphoma), or at least unclassified clonal lymphoproliferation or reactive lymphocytosis [20, 21].
These findings account for the rationale of treating refractory and
severe CAD cases with rituximab [30], alone or in combination
with fludarabine [31] or bendamustine, particularly when associated with chronic lymphocytic leukemia [32], or with bortezomib
[33]. Since most RBC destruction does not occur in the spleen, it is
not surprising that splenectomy is usually ineffective in CAD
[2022].

Transfus Med Hemother 2015;42:287293

289

Fig. 1. T-lymphocyte subsets and cytokine interactions in AIHA. Cytokines that were found elevated are highlighted in yellow: IL-2 and IL-12,
that induce the differentiation of CD4+ nave
T cells into Th1 subset, and IL-4, that promotes
Th2 switch. Elevated levels of TGF- favor the
differentiation of Th17 subset, which amplifies the
pro-inflammatory and autoimmune response. On
the contrary, decreased levels of both IFN- and
Tregs were found (highlighted in green). The
former resulted in decreased inhibition of Th2
response, i.e. an amplification of the autoantibodymediated autoimmune disease, and the latter may
cause a lack of down-regulation of inflammatory
and autoimmune pathways.

T Lymphocytes and Immunoregulatory Cytokines in


the Pathogenesis of AIHA
The T-cell mediated regulation of the humoral immune system
has been shown to play a critical role in loss of self-tolerance in
AIHA. In the past years increased in vitro proliferative response of
T cells and increased cytotoxicity of natural killer cells against autologous RBCs have been described in AIHA [34, 35]. Until a few
years ago, CD4+ T helper cells were subdivided into two major Tcell subsets, termed the Th1 and Th2 subsets. More recently, Th17
cells, which are characterized by secretion of IL-17, have been
identified as key effectors in the development of many autoimmune diseases, including AIHA [40]. Elevated frequency of Th17
cells and increased IL-17 secretion were found closely correlated
with the disease activity in AIHA patients. Consistently, in the rat
model of AIHA, the adoptive transfer of Th17 cells heightened
anti-RBC antibody responses and increased the onset of AIHA,
whereas disease onset was abrogated by in vivo neutralization of
IL-17 [36].
Moreover, CD4+ CD25+ Tregs are now recognized as key players in immunologic tolerance and, thereby, in preventing AIHA
and other polyclonal autoimmune disorders. Ward et al. [37] firstly
described autoantigen-specific, IL-10-secreting Treg clones recovered ex vivo from a patient with AIHA. These clones recognized a
peptide, 72H-86L, derived from the Rh RBC autoantigen and expressed different T regulatory markers depending on activation
state: high levels of CD25 and LAG-3 when expanding nonspecifically, but FoxP3 after activation by the autoantigen they recognize
[38]. More recently, circulating CD4+ Tregs were found reduced in
patients with AIHA, along with high levels of IL-10 and IL-12 [39].

290

Transfus Med Hemother 2015;42:287293

It is known that Tregs produce IL-10 and transforming growth factor (TGF-), and the IL-10/IL-12 imbalance may thus play an essential role in the onset and/or maintenance of AIHA. Several
other abnormalities of immunoregulatory cytokines have been reported in AIHA: IL-1, IL-2/IL-2R, IL-6, and IL-21 serum levels
were found increased; in culture experiments, the Th1 cytokines
IL-2 and IL-12 were reported elevated, whereas IFN- was found
reduced. Moreover, the Th2 cytokines IL-4 and IL-13 were reported increased, together with elevated production of IL-6, IL-10,
and IL-17 [36, 40, 41]. Patients with active hemolysis showed further reduction of IFN- and increased secretion of TGF-. Moreover, addition of IL-4, IL-6, IL-10, IL-13, and TGF- increased autoantibody secretion in culture [40]. All these data are in favor of a
reduced Th1- and a predominant Th2-like profile in the immunopathogenesis of AIHA, in contrast to the prevalent role of CD8+
IFN--secreting cytotoxic T cells in organ-specific autoimmunity
(i.e multiple sclerosis, experimental autoimmune encephalomyelitis, type 1 diabetes, and autoimmune thyroiditis). Moreover, elevated levels of TGF- favor the differentiation of a Th17 subset,
which amplifies the pro-inflammatory and autoimmune response.
On the contrary, decreased levels of both IFN- and Tregs resulted
in decreased inhibition of Th2 response, i.e. an amplification of the
autoantibody-mediated autoimmune disease (fig. 1). Notwithstanding these cytokine abnormalities, no difference was reported
in the phenotypic distributions of polymorphisms of the TNF-,
IL-10, IL-12, and CTLA-4 between AIHA patients and controls. As
regards therapy, immunoregulatory cytokines were evaluated after
low-dose rituximab (100 mg fixed dose for 4 weekly infusions)
[42]. The clinical response was correlated with an increase of IFN, and a reduction of IL-4 and IL-17, suggesting that the drug ex-

Barcellini

Table 2. Main immunological abnormalities


involved in AIHA pathogenesis

Pathogenic mechanism
Autoantibodies
IgG (all subclasses)
IgG1 and IgG3
IgM

antibody-dependent cellular cytotoxicity (ADCC)


complement activation
strong complement activation

Complement system

membrane attack complex (MAC)


phagocytosis of C3b-opsonized erythrocytes

Cellular immunity

increased cytotoxicity of natural killer cells


activated autoreactive CD4+ T helper 1
increased number of Th17 cells
reduced peripheral CD4+ T-reg

Immunoregulatory cytokines

high serum levels of IL-1, IL-2/IL-2R, IL-6, IL-21


increased production in culture of T helper 1 cytokine IL-2 and IL-12
reduced production of IFN- in culture
high levels of T helper 2 cytokines IL-4 and IL-13 in culture
increased production in culture of IL-6 and IL-10
elevated levels of IL-17 in culture
increased production in culture of TGF- in active patients

erts an immunomodulating activity together with the well-known


B-depleting effect. Finally, recent unpublished observations report
increased serum values of BAFF and APRIL in AIHA, opening
future therapeutic options with monoclonal antibodies directed
against this factors, e.g. belimumab. Table2 summarizes the main
immunological abnormalities involved in AIHA pathogenesis.

short-lived into long-lived plasma cells. The authors conclude that


the presence of splenic long-lived autoreactive plasma cells may explain why some wAIHA patients cannot achieve a response after
rituximab. The B-cell depletion induced by rituximab itself promotes a suitable environment for the maturation of autoimmune
long-lived plasma cells in the spleen. Targeting specifically some
factors such as BAFF right after rituximab injection might be an
interesting therapeutic option in the future.

Role of the Spleen in the Pathogenesis of AIHA


The spleen is a major organ for antibody synthesis and immune
destruction of IgG-coated erythrocytes. This is the reason why it is
considered the most effective conventional second-line treatment
of warm AIHA [43]. However, splenectomy is ineffective in about
30% of cases, presumably because there is still an elevated synthesis
of autoantibodies in other lymphoid organs, and the liver replaces
the spleens function of RBC destruction. Unfortunately, the extent
of splenic sequestration assessed by radiolabeled erythrocytes as
well as the disease duration and the response to steroids are not
reliable predictors of its effectiveness [44]. The rate of splenectomy
is about 1015% of all AIHA cases but is nowadays decreasing
given the availability of equally effective second-line medical treatments, primarily rituximab. It is worth reminding that splenectomy is usually ineffective in CAD since most RBC destruction occurs in the liver [20, 21]. An interesting recently reported finding is
the emergence of long-lived autoreactive plasma cells in the spleen
of primary wAIHA patients treated with rituximab [45]. The
spleen was obtained after surgery for relapse or failure of rituximab
and analyzed by microscopy and flow cytometry, showing the presence of Bcl6+ germinal-center B cells. The residual population of
CD19+ B cells includes non-proliferative memory B cells and
plasma cells, which secreted anti-RBC IgG in vitro. Furthermore,
the cytokine BAFF was increased in the supernatants of spleen cell
cultures, suggesting that it may play a role in the differentiation of

New Insights in the Pathogenesis of Autoimmune


Hemolytic Anemia

Additional Factors That Influence the Clinical


Characteristics of AIHA
The major determinants of the clinical severity of AIHA are the
autoantibody pathogenicity (related to their class, subclass, thermal
amplitude as well as affinity and efficiency in activating complement) and the activity of the reticuloendothelial system. However,
there are other factors that influence the clinical picture of AIHA,
in particular the efficacy of erythroblastic compensatory response,
represented by the absolute number of reticulocytes in the peripheral blood. Reticulocytopenia is not rare in AIHA, particularly in
children and in severe cases [4648], and may be due to autoimmune reaction against bone marrow erythroid precursors. There is
emerging evidence that the administration of erythropoietin may
be useful to overcome the temporary bone marrow failure in compensating hemolysis [49], which is in line with the efficacy of
thrombopoietin agonists in primary immune thrombocytopenia
[50]. Moreover, the use of erythropoietin may reduce transfusion
need and avoid hemolysis related to overtransfusion.
The relative weight of all factors involved in the pathogenesis of
AIHA could account for the great heterogeneity of the disease. A
recent large multicenter study of 308 AIHA cases showed that 60%
were wAIHA, 27% CAD, 8% mixed, and 5% atypical (mostly DATnegative). A severe presentation (Hb levels < 6 g/dl) was observed

Transfus Med Hemother 2015;42:287293

291

in about one-third of cases, mainly warm IgG+C, mixed and atypical forms, with frequently associated reticulocytopenia [10]. As regards therapy, the majority of wAIHA patients received first-line
steroid therapy only, whereas patients with mixed and atypical
forms had an increased risk of relapse and were more frequently
treated with 2 or more therapy lines. Thrombotic events were associated with a severe onset, intravascular hemolysis and previous
splenectomy, and predictors of a fatal outcome were severe infections, particularly in splenectomized cases, acute renal failure,
Evans syndrome, and multi-treatment (4 or more lines) [10].
Moreover, we recently described 13 acute and very severe cases
of AIHA, displaying multi-drug refractoriness and related complications, of whom 7 were fatal. These challenging AIHAs showed a
highly unpredictable clinical courses, suggesting that rapidly evolving cases deserve a scrupulous attention by experienced clinicians
to avoid a delay in potentially life-saving strategies. In particular,
the presence of intravascular hemolysis, hemoglobinuria, DAT
positivity for IgG+C, reticulocytopenia, and/or Evans syndrome
should alarm clinicians to generously give steroid boluses, to early
utilize rituximab, and to consider plasma exchange in critical conditions. We also suggest performing splenectomy soon after rituximab failure, because of the poor prognosis of surgery after multiple
immunosuppressive therapies. Moreover, we suggest the use of
erythropoietin to reduce overtransfusion and an anti-thrombotic
prophylaxis, particularly in splenectomized patients [11]. Currently we use low-molecular-weight heparins at prophylactic doses,
i.e. 3050% of the anticoagulant dose, as long as the hemolytic process is active [51].
An additional factor that influence the clinical picture of AIHA
is the association with lymphoproliferative diseases. It is known
that about 10% of patients with chronic lymphocytic leukemia and
23% of those with non-Hodgkins and Hodgkins lymphoma develop AIHA. Moreover, in a population-based study of 86 patients
with CAD, a clonal bone marrow lymphoproliferation was demonstrated in 75% and 90% of the patients by bone marrow biopsy and
flow cytometry, respectively [20]. Moreover, there is increasing evidence that severe and fatal AIHA may be a complication of hematopoietic stem cell transplantation. A recent large study at Kings
College Hospital [52] described an overall incidence of 3.6% and
an association with hematopoietic stem cell transplantation from
unrelated donors. About two-thirds of cases required multiple
treatment, but only half achieved complete resolution of AIHA.
Ten cases were fatal, and death was directly attributable to AIHA
in 4 cases, indicating that post-transplantation AIHA had a higher
overall mortality (HR 2.48) than primary AIHA.

Finally, it is worth reminding the passenger lymphocyte syndrome, which is due to the production of antibodies directed
against recipient RBCs by passively transferred lymphocytes transfused with the stem cell, most often observed when the donor is
group O and the patient group A. A transient immune hemolysis
has also been associated with solid organ transplantation, with a
risk and degree of hemolysis in proportion to the mass of lymphocytes transplanted being lowest in kidney (antibody in 17%, hemolysis in 9%), next higher in liver (antibody in 40%, hemolysis in
29%), and highest in heart-lung transplants (antibody in 70%, hemolysis in 70%) [53, 54].

Conclusions
AIHA is an heterogeneous condition including warm, cold, and
mixed forms, along with atypical cases of difficult diagnostic classification (DAT-negative, warm IgM), which are reported with increasing frequency. It can be primary or secondary to lymphoproliferative syndromes, infections, immunodeficiency, and tumors.
Moreover, AIHA is described with increasing frequency following
hematopoietic stem cell transplantation, and in recipients of solid
organ transplants. The clinical presentation of primary AIHA may
vary from mild to very severe forms, with a reported mortality of
11% in older series, and of 4% in a more recent studies; secondary
forms are usually more severe, with an increased mortality, particularly in post-transplant forms. Several immunologic mechanisms are involved in the pathogenesis of AIHA: autoantibodies,
ADCC, phagocytes, B and T lymphocytes, Tregs, cytokines, and
the complement system. New insights in this complex scenario
may give hints for future therapeutic options specifically aimed at
correcting the main pathogenic mechanism involved in the different forms of AIHA.

Acknowledgement
This work was supported by research funding from Fondazione IRCCS Ca
Granda Ospedale Maggiore Policlinico, grant number RC 2014 and by Ministry
of Health, grant number RF 2010 convention N. 141/RF-20102303934.

Disclosure Statement
The author has no conflict of interest to declare.

References
1 Petz LD, Garratty G: Immune Hemolytic Anemias,
2nd ed. Philadelphia, Churchill Livingstone, 2004.
2 Berentsen S: How I manage cold agglutinin disease. Br
J Haematol 2011; 153: 309317.
3 Gehrs BC, Friedberg RC: Autoimmune hemolytic anemia. Am J Hematol 2002; 69: 258271.

292

4 Abbas AK, Lichtman AH, Pillai S: Cellular and Molecular Immunology, Philadelphia, Saunders, 2012.
5 McQueen F: A B cell explanation for autoimmune disease: the forbidden clone returns. Postgrad Med J.
2012; 88: 226233.

Transfus Med Hemother 2015;42:287293

6 Garratty G, Arndt PA: Drugs that have been shown to


cause drug-induced immune hemolytic anemia or positive direct antiglobulin tests: some interesting findings
since 2007. Immunohematology 2014; 30: 6679.
7 Salama A: Drug-induced immune hemolytic anemia.
Expert Opin Drug Saf 2009; 8: 7379.

Barcellini

8 Ahrens N, Pruss A, Khne A, Kiesewetter H, Salama A:


Coexistence of autoantibodies and alloantibodies to
red blood cells due to blood transfusion. Transfusion
2007; 47: 813816.
9 Alijotas-Reig J: Human adjuvant-related syndrome or
autoimmune/inflammatory syndrome induced by adjuvants. Where have we come from? Where are we
going? A proposal for new diagnostic criteria. Lupus
2015; 2015, doi: 10.1177/0961203315579092.
10 Barcellini W, Fattizzo B, Zaninoni A, Radice T, Nichele
I, Di Bona E, Lunghi M, Tassinari C, Alfinito F, Ferrari
A, Leporace AP, Niscola P, Carpenedo M, Boschetti C,
Revelli N, Villa MA, Consonni D, Scaramucci L, De
Fabritiis P, Tagariello G, Gaidano G, Rodeghiero F,
Cortelezzi A, Zanella A: Clinical heterogeneity and
predictors of outcome in primary autoimmune hemolytic anemia: a GIMEMA study of 308 patients. Blood
2014; 124: 29302936.
11 Fattizzo B, Zaninoni A, Nesa F, Sciumbata VM,
Zanella A, Cortelezzi A, Barcellini W: Lessons from
very severe, refractory and fatal primary autoimmune
hemolytic anemias. Am J Hematol 2015; 90:E149151.
12 Arndt PA, Leger RM, Garratty G: Serologic findings in
autoimmune hemolytic anemia associated with immunoglobulin M warm autoantibodies Transfusion 2009;
49: 235242.
13 Bartolmas T, Salama A: A dual antiglobulin test for the
detection of weak or nonagglutinating immunoglobulin M warm autoantibodies. Transfusion 2010; 50:
11311134.
14 Bass GF, Tuscano ET, Tuscano JM: Diagnosis and classification of autoimmune hemolytic anemia. Autoimmun Rev 2014; 13: 560564.
15 Garratty G: Immune hemolytic anemia caused by
drugs. Expert Opin Drug Saf 2012; 11: 635642.
16 Packman CH: Hemolytic anemia due to warm autoantibodies. Blood Rev 2008; 22: 1731.
17 Dacie J: Auto-immune haemolytic anaemia [AIHA]:
pathogenesis; in Dacie J (ed): The Haemolytic Anaemias. Vol. 3. London, Churchill Livingstone, 1992, pp.
392451.
18 Kurlander RJ, Rosse WF, Logue GL: Quantitative influence of antibody and complement coating of red
cells on monocyte-mediated cell lysis. J Clin Invest
1978; 61: 13091319.
19 Berentsen S, Beiske K, Tjnnfjord GE: Primary chronic
cold agglutinin disease: an update on pathogenesis,
clinical features and therapy. Hematology 2007; 12:
361370.
20 Berentsen S, Ulvestad E, Langholm R, Beiske K,
Hjorth-Hansen H, Ghanima W, Srb JH, Tjnnfjord
GE: Primary chronic cold agglutinin disease: a population based clinical study of 86 patients. Haematologica
2006; 91: 460466.
21 Swiecicki PL, Hegerova LT, Gertz MA: Cold agglutinin
disease. Blood 2013; 122: 11141121.
22 Ulvestad E, Berentsen S, Mollnes TE : Acute phase haemolysis in chronic cold agglutinin disease. Scand J Immunol 2001; 54: 239242.
23 Roth A, Huttmann A, Rother RP, Duhrsen U, Philipp
T: Long-term efficacy of the complement inhibitor
eculizumab in cold agglutinin disease. Blood 2009; 113:
38853886
24 Hill A, Rother RP, Arnold L, Kelly R, Cullen MJ, Richards SJ, Hillmen P: Eculizumab prevents intravascular
hemolysis in patients with paroxysmal nocturnal
hemoglobinuria and unmasks low-level extravascular
hemolysis occurring through C3 opsonization. Haematologica 2010; 95: 567573.
25 Shi J, Rose EL, Singh A, Hussain S, Stagliano NE, Parry
GC, Panicker S: TNT003, an inhibitor of the serine
protease C1s, prevents complement activation induced
by cold agglutinins. Blood 2014; 123: 40154022.

New Insights in the Pathogenesis of Autoimmune


Hemolytic Anemia

26 Wouters D, Stephan F, Strengers P, de Haas M, Brouwer C, Hagenbeek A, van Oers MH, Zeerleder S: C1esterase inhibitor concentrate rescues erythrocytes
from complement-mediated destruction in autoimmune hemolytic anemia. Blood 2013; 121: 12421244.
27 Risitano AM, Notaro R, Pascariello C, Sica M, del Vecchio L, Horvath CJ, Fridkis-Hareli M, Selleri C, Lindorfer MA, Taylor RP, Luzzatto L, Holers VM: The
complement receptor 2/factor H fusion protein TT30
protects paroxysmal nocturnal hemoglobinuria erythrocytes from complement-mediated hemolysis and C3
fragment. Blood 2012; 119: 63076316.
28 Risitano AM1, Ricklin D, Huang Y, Reis ES, Chen H,
Ricci P, Lin Z, Pascariello C, Raia M, Sica M, Del
Vecchio L, Pane F, Lupu F, Notaro R, Resuello RR,
DeAngelis RA, Lambris JD: Peptide inhibitors of C3
activation as a novel strategy of complement inhibition
for the treatment of paroxysmal nocturnal hemoglobinuria. Blood 2014; 123: 20942101.
29 Harboe M, van Furth R, Schubothe H, Lind K, Evans
RS: Exclusive occurrence of K chains in isolated cold
haemagglutinins. Scand J Haematol 1965: 2:259266.
30 Berentsen S, Tjnnfjord GE: Diagnosis and treatment
of cold agglutinin mediated autoimmune hemolytic
anemia. Blood Rev 2012; 26: 107115.
31 Berentsen S, Randen U, Vgan AM, Hjorth-Hansen H,
Vik A, Dalgaard J, Jacobsen EM, Thoresen AS, Beiske
K, Tjnnfjord GE: High response rate and durable remissions following fludarabine and rituximab combination therapy for chronic cold agglutinin disease.
Blood 2010; 116: 31803184.
32 Quinquenel A, Willekens C, Dupuis J, Royer B, Ysebaert L, De Guibert S, Michallet AS, Feugier P, Guieze
R, Levy V, Delmer A: Bendamustine and rituximab
combination in the management of chronic lymphocytic leukemia-associated autoimmune hemolytic anemia: a multicentric retrospective study of the French
CLL intergroup (GCFLLC/MW and GOELAMS). Am
J Hematol 2015; 90: 204207.
33 Carson KR, Beckwith LG, Mehta J: Successful treatment of IgM-mediated autoimmune hemolytic anemia
with bortezomib. Blood 2010; 115: 915.
34 Fagiolo E: Immunological tolerance loss vs. erythrocyte self antigens and cytokine network disregulation
in autoimmune hemolytic anaemia. Autoimmun Rev
2004; 3: 5359.
35 Gilsanz F, de La Serna J, Molt L, Alvarez-Mon M:
Hemolytic anemia in chronic large granular lymphocytic leukemia of natural killer cells: cytotoxicity of
natural killer cells against autologous red cells is associated with hemolysis. Transfusion 1996; 36: 463466.
36 Xu L, Zhang T, Liu Z, Li Q, Xu Z, Ren T: Critical role
of Th17 cells in development of autoimmune hemolytic anemia. Exp Hematol 2012; 40: 9941004.
37 Ward FJ, Hall AM, Cairns LS, Leggat AS, Urbaniak SJ,
Vickers MA, Barker RN: Clonal regulatory T cells specific for a red blood cell autoantigen in human autoimmune hemolytic anemia. Blood 2008; 111: 680687.
38 Sakaguchi S, Ono M, Setoguchi R, Yagi H, Hori S, Fehervari Z, Shimizu J, Takahashi T, Nomura T: Foxp3+
CD25+ CD4+ natural regulatory T cells in dominant
self-tolerance and autoimmune disease. Immunol Rev
2006; 212: 827.
39 Ahmad E, Elgohary T, Ibrahim H: Naturally occurring
regulatory T cells and interleukins 10 and 12 in the
pathogenesis of idiopathic warm autoimmune hemolytic anemia. J Investig Allergol Clin Immunol 2011;
21: 297304.
40 Barcellini W, Clerici G, Montesano R, Taioli E, Morelati F, Rebulla P, Zanella A: In vitro quantification of
anti-red blood cell antibody production in idiopathic
autoimmune haemolytic anaemia: effect of mitogen
and cytokine stimulation. Br J Haematol 2000; 111:
452460.

41 Toriani-Terenzi C, Fagiolo E: IL-10 and the cytokine


network in the pathogenesis of human autoimmune
hemolytic anemia. Ann N Y Acad Sci 2005;1051: 2944.
42 Barcellini W, Zaja F, Zaninoni A, Imperiali FG, Battista ML, Di Bona E, Fattizzo B, Consonni D, Cortelezzi A, Fanin R, Zanella A: Low-dose rituximab in
adult patients with idiopathic autoimmune hemolytic
anemia: clinical efficacy and biologic studies. Blood
2012; 119: 36913697.
43 Lechner K, U Jager U: How I treat autoimmune hemolytic anemias in adults. Blood 2010; 16: 18311838.
44 Crowther M, Chan YL, Garbett IK, Lim W, Vickers
MA, Crowther MA: Evidence-based focused review of
the treatment of idiopathic warm immune hemolytic
anemia in adults. Blood 2011; 118: 40364040.
45 Mahvas M, Michel M, Vingert B, Moroch J, Boutboul
D, Audia S, Cagnard N, Ripa J, Menard C, Tarte K,
Mgret J, Le Gallou S, Patin P, Thai L, Galicier L, Bonnotte B, Godeau B, Noizat-Pirenne F, Weill JC, Reynaud CA: Emergence of long-lived autoreactive plasma
cells in the spleen of primary warm auto-immune
hemolytic anemia patients treated with rituximab. J
Autoimmun 2015; doi: 10.1016/j.jaut.2015.05.006.
46 Conley CL, Lippman SM, Ness P: Autoimmune hemolytic anemia with reticulocytopenia. A medical emergency. JAMA 1980; 244: 16881690.
47 Liesveld JL, Rowe JM, Lichtman MA: Variability of the
erythrocyte response in autoimmune hemolytic anemias: analysis of 109 cases. Blood 1987; 69: 820826.
48 Aladjidi N, Leverger G, Leblanc T, Picat MQ, Michel G,
Bertrand Y, Bader-Meunier B, Robert A, Nelken B,
Gandemer V, Savel H, Stephan JL, Fouyssac F, Jeanpetit J, Thomas C, Rohrlich P, Baruchel A, Fischer A,
Chne G, Perel Y; Centre de Rfrence National des
Cytopnies Auto-immunes de lEnfant (CEREVANCE): New insights into childhood autoimmune
hemolytic anemia: a French national observational
study of 265 children. Haematologica 2011;96:655663.
49 Arbach O, Funck R, Seibt F, Salama A: Erythropoietin
may improve anemia in patients with autoimmune
hemolytic anemia associated with reticulocytopenia.
Transfus Med Hemother 2012; 39: 221223.
50 Kuter DJ: The biology of thrombopoietin and thrombopoietin receptor agonists. Int J Hematol 2013; 98:
1023.
51 Akl EA, Ramly EP, Kahale LA, Yosuico VE, Barba M,
Sperati F, Cook D, Schnemann H: Anticoagulation
for people with cancer and central venous catheters.
Cochrane Database Syst Rev 2014; 10:CD006468.
52 Wang M, Wang W, Abeywardane A, Adikarama M,
McLornan D, Raj K, de Lavallade H, Devereux S, Mufti
GJ, Pagliuca A, Potter VT, Mijovic A: Autoimmune
hemolytic anemia after allogeneic hematopoietic stem
cell transplantation: analysis of 533 adult patients who
underwent transplantation at Kings College Hospital.
Biol Blood Marrow Transplant 2015; 21: 6066.
53 Hoffman PC: Immune hemolytic anemia selected
topics. Hematology Am Soc Hematol Educ Program
2009: 8086.
54 ElAnsary M, Hanna MO, Saadi G, ElShazly M, Fadel
FI, Ahmed HA, Aziz AM, ElSharnouby A, Kandeel
MM: Passenger lymphocyte syndrome in ABO and
Rhesus D minor mismatched liver and kidney transplantation: A prospective analysis. Hum Immunol
2015; 76: 447452.

Transfus Med Hemother 2015;42:287293

293

Vous aimerez peut-être aussi